Synthesis and Evaluation of Potent and Selective β3 Adrenergic

(b) Himms-Hagen, J.; Melnyk, A.; Zingaretti, M. C.; Ceresi, E.; Barbatelli, G.; Cinti, S. Multiocular fat cells in WAT of CL-316,243-treated rats deri...
0 downloads 0 Views 324KB Size
J. Med. Chem. 2002, 45, 567-583

567

Articles Synthesis and Evaluation of Potent and Selective β3 Adrenergic Receptor Agonists Containing Acylsulfonamide, Sulfonylsulfonamide, and Sulfonylurea Carboxylic Acid Isosteres† David E. Uehling,*,‡ Kelly H. Donaldson,‡ David N. Deaton,‡ Clifton E. Hyman,‡ Elizabeth E. Sugg,‡ David G. Barrett,‡ Robert G. Hughes,‡ Barbara Reitter,‡ Kim K. Adkison,§ Mary E. Lancaster,| Frank Lee,§ Robert Hart,§ Mark A. Paulik,| Bryan W. Sherman,⊥ Timothy True,⊥ and Conrad Cowan⊥ Departments of Medicinal Chemistry, Receptor Biology, Research Bioanalysis and Drug Metabolism, and Pharmacology, GlaxoSmithKline, Research Triangle Park, North Carolina 27709 Received April 2, 2001

Starting from phenethanolamine aniline leads 3a and 3b, we have identified a series of functionally potent and selective β3 adrenergic receptor (AR) agonists containing acylsulfonamide, sulfonylsulfonamide, or sulfonylurea groups within the aniline phenethanolamine series. In β3, β2, and β1 AR cAMP functional assays, 3a and other right-hand side (RHS) carboxylate analogues were found to be full agonists that were modestly selective against β1 or β2 ARs, while analogues lacking RHS acid functionality were active at β3 AR but not selective. Replacement of the carboxylate with acylthiazole and acylmethylsulfone gave potent, but only modestly selective, compounds. Increasing the size of the RHS sulfonamide substituent with phenyl or p-toluene afforded compounds with good potency and functional selectivity (β3 AR pEC50 greater than 8; β1 and β2 AR selectivity greater than 40- and 500-fold, respectively). Our SAR studies suggest that the potency and selectivity profile of the best analogues reported here is a result of both the steric bulk and acidity of the RHS sulfonamide NH group. Although all of the analogues had a pharmacokinetic half-life of less than 2 h, acylsulfonamides 43 and 44 did show moderately low clearance in dogs. These two compounds were further evaluated by thermographic imaging in mice and were found to produce a robust thermogenic response via oral administration. Introduction The increasing prevalence of obesity and its associated comorbidities in industrialized nations, including type 2 diabetes mellitus and related cardiovascular disorders, has stimulated efforts to develop effective new approaches in the treatment of this condition. While most therapeutic approaches involve altering the balance of metabolic energy by reducing energy intake, an alternative approach for the management of obesity is to effect an increase in the rate of energy expenditure (thermogenesis).1 In 1984, compounds of the phenethanolamine class having thermogenic properties in rodents were first disclosed as potential agents in the treatment of human obesity.2 Despite their structural similarity to known β1 and β2 adrenoceptor (AR) ligands, pharmacological studies indicated that these compounds stimulate a third, or “atypical”, β AR that is now described as “β3” AR.3 Although this receptor was not characterized for several years, the striking rodent pharmacology of early phenethanolamine compounds †

Dedicated to the memory of our colleague Barbara Reitter. * To whom correpsondence should be addressed. Phone: 919-4836244. Fax: 919-483-6053. E-mail: [email protected]. ‡ Department of Medicinal Chemistry. § Department of Research Bioanalysis and Drug Metabolism. | Department of Pharmacology. ⊥ Department of Receptor Biology.

triggered considerable activity at a number of pharmaceutical companies in the search for new antidiabetic and antiobesity drugs.4 Before the cloned human receptor became available, early efforts focused on optimization using rodent models and led to the identification of clinical candidates such as 15 and 2.6 Unfortunately, although some indication of efficacy was seen in clinical studies with these “first generation” development candidates, these and other early β3 AR agonists have not advanced beyond phase II clinical trials because of lack of efficacy, poor pharmacokinetics, or dose-limiting cardiovascular side effects.7,8 Despite the failure of early β3 agonists as viable therapeutic agents, in the past 12 years additional evidence has accumulated to suggest that β3 AR stimulation may have potential benefit in the treatment of diabetes and obesity. In 1989, the human,9 rat,10 and mouse11 β3 ARs were first cloned and characterized. Evaluation of the activity of numerous compounds on the cloned rodent and human receptors uncovered significant interspecies differences in their activities at the three β AR subtypes.12 These differences may, in part, explain the poor efficacy and/or side effect profile of early clinical candidates.7 Adding support to the validity of the β3 AR as an important therapeutic target, rodent studies have suggested a “reawakening” of

10.1021/jm0101500 CCC: $22.00 © 2002 American Chemical Society Published on Web 12/29/2001

568

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3

dormant β3 AR responsive brown adipose tissue upon prolonged stimulation with 2.13 Recent studies in nonhuman primates treated with a selective β3 AR agonist have demonstrated a separation in the dose response between metabolic rate and cardiovascular response, along with upregulation of uncoupling protein in axillary fat indicative of increased thermogenic potential.14 In terms of human validation, β3 ARs have been detected in human tissues of possible relevance to the treatment of type 2 diabetes and obesity, including fat and skeletal muscle.15,16 In human clinical studies, significant increases in resting metabolic rate were seen using isoproterenol or 1 that were in part resistant to the β1 AR and β2 AR blocking effects of nadolol, implying that β3 AR agonism plays a role in mediating human energy expenditure.7 In other studies, compound 2, reported to have minimal β1 AR and β2 AR activity,6,12 caused increased insulin sensitivity and glucose utilization in lean, healthy volunteers.8 Finally, it has been proposed that a Trp64Arg β3 AR mutation in the human population plays a role in the development of diabetes and/or obesity in some individuals possessing this genetic variant.17 In the past decade, drug discovery efforts have shifted toward use of cell lines expressing human receptors in primary screening assays in order to identify potent β3 AR agonists that are selective over human β2 and β1 ARs. Several structure-activity relationship (SAR) studies have now been published that describe compounds characterized in human β3, β2, and β1 AR expressing cell lines. With the exception of two articles reporting tetrahydroisoquinoline β3 AR agonists,18a,b these papers have described compounds in either the arylethanolamine or aryloxypropanolamine class. Among these analogues, a great deal of structural diversity has appeared in the right-hand side (RHS) portion of these analogues distal to the arylethanolamine or aryloxypropanolamine pharmacophore, suggesting a permissive interaction with the receptor in this portion of the molecule.19 For example, in a series of papers by workers at Merck, phenylsulfonamides bearing a variety of RHS phenyl ring substituents have been described. A representative example of this series is the thiazole phenylsulfonamide 4 that has been described as a phase I clinical candidate.20 In another article, a laboratory at Bristol-Myers Squibb (BMS) has reported the SAR of phenethanolamines related to 1 with modified RHS ether substituents. Consistent with earlier SAR studies in rodent systems,3,21 the BMS work highlighted an important role of carboxylate and other negatively charged groups (such as sulfonic acid 5) in providing β1 and β2 AR selectivity within certain phenethanolamine analogues22a (see Chart 1 for structures of some β3 agonists). Also consistent with these results, a recent paper has described the use of thiazolidinediones as acid isostere replacements in both arylethanolamine and aryloxypropanolamine β3 AR agonists.22b In 1995, a Glaxo patent disclosed a series of phenethanolamines containing an anilinophenylacetic acid RHS subunit, as exemplified by GR9803 (3a).23 Although it is a potent (95% R,R diastereomer. 1H NMR (400 MHZ, CDCl3): δ -0.15 (s, 3H), 0.05 (s, 3H), 0.95 (s, 9H), 1.25 (d, 3H, J ) 7 Hz), 2.62-2.87 (m, 2H), 3.38 (q, 1H, J ) 7 Hz), 3.67 (s, 3H), 4.65-4.75 (d, 1H), 7.18-7.23 (m, 3H), 7.25 (m, 1H). Methyl (2R)-2-{(tert-Butoxycarbonyl)[(2R)-2-{[tert-butyl(dimethyl)silyl]oxy}-2-(3-chlorophenyl)ethyl]amino}propanoate (9).23 The amine 8 (22.4 g, 60.2 mmol) and ditert-butyl dicarbonate (14.3 g, 65.5 mmol) were combined and heated neat in an oil bath at 90-95 °C for 5 min. (Caution: a blast shield should be used when heating this mixture). After the mixture was cooled, additional di-tert-butyl dicarbonate (2.73 g, 12.5 mmol) was added and the mixture was heated at 95 °C for 10 min. The mixture was allowed to cool to ambient temperature and was purified by silica gel chromatography (eluting with 10:1 hexane/EtOAc followed by 5:1 hexane/ EtOAc) to supply 27.1 g (96%) of Boc amine 9 as a colorless oil, which 1H NMR indicated to be a 60:40 mixture of rotamers. 1H NMR (400, CDCl ): δ 0.06 (s, 3H), 0.89 (s, 9H), 1.09 (d, 3 1.25 H, J ) 7.2 Hz), 1.31 (d, 1.75H, J ) 6.8 Hz), 1.41 (s, 3.6 H), 1.43 (s, 5.4 H), 3.10-3.20 (m, 1H), 3.35 (d, 1H, J ) 6.0 Hz), 3.40-3.50 (m, 1H), 3.69 (s, 3H), 3.82-3.93 (m, 0.5 H),

Uehling et al. 4.05-4.15 (m, 0.5H), 4.82-4.95 (m, 0.5 H), 5.00-5.05 (m, 0.5 H), 7.10-7.30 (m, 3H), 7.35 (s, 0.5 H), 7.40 (s, 0.5H). tert-Butyl (2R)-2-{[tert-Butyl(dimethyl)silyl]oxy}-2-(3chlorophenyl)ethyl[(1R)-1-methyl-2-oxoethyl]carbamate (10).23 To a solution of the methyl ester 9 (3.83 g, 8.15 mmol) in anhydrous toluene (40 mL) at -78 °C was added 1.5 M DIBAL in toluene (13.5 mL, 20.3 mmol) dropwise over 2 min. The mixture was stirred at -78 °C for 1 h and quenched by slow addition of MeOH (3 mL) followed by 15% w/v potassium sodium tartrate (20 mL). The resulting mixture was allowed to warm to ambient temperature, stirred vigorously for 1 h, and filtered through a pad of Celite. After the mixture was washed with additional EtOAc, the filtrate was partitioned between water (50 mL) and EtOAc (50 mL). The organic layer was dried, filtered, and concentrated to afford the product aldehyde 10 (3.43 g, 96%) as a colorless oil judged by 1H NMR integration of aldehyde peaks to be a 13:1 mixture of R,R/ minor diastereomers. 1H NMR (400 MHz, CDCl3): δ -0.10 (s, 3H), 0.01 (s, 1.5 H), 0.02 (s, 1.5H), 0.85 (s, 9H), 1.20-1.25 (m, 3H), 1.38 (s, 4.5H), 1.40 (s, 4.5H), 3.0-3.15 (m, 1H), 3.253.40 (m, 1H), 3.3.70-3.85 (m, 1H), 4.90 (t, 0.5H), 5.02 (t, 0.5H), 7.18-7.25 (m, 3H), 7.28 (s, 0.5H), 7.35 (s, 0.5H), 9.20 (s, 0.5H), 9.38 (s, 0.5H). N-[2-(4-Nitrophenyl)ethyl]-4-methylbenzenesulfonamide.25 An N2-purged flask equipped with a stirring bar was charged with 2-(4-nitrophenyl)ethylamine hydrochloride (2.0 g, 9.87 mmol), p-toluenesulfonyl chloride (1.88 g, 9.87 mmol), and anhydrous CH2Cl2 (50 mL). Triethylamine (1.51 mL, 10.86 mmol) was added, and the resulting mixture was stirred at ambient temperature for 20 h, diluted with CH2Cl2 (25 mL), and washed with 1.0 N aqueous HCl (25 mL). The organic layer was separated, washed with saturated aqueous NaHCO3 (25 mL), dried, filtered, and concentrated to give a light-yellow solid. Trituration with MeOH afforded a solid that was washed with ether to supply 1.04 g (33% yield) of the product as a white solid. 1H NMR (DMSO-d6): δ 2.47 (s, 3H), 2.94 (t, 2H, J ) 6.9 Hz), 3.31 (q, 2H, J ) 6.9 Hz), 4.51 (t, 1H, J ) 6.3 Hz), 7.29-7.34 (m, 4H), 7.73 (d, 2H, J ) 8.1 Hz), 8.15 (d, 2H, J ) 8.4 Hz). MS (ES), m/e (M - H) ) 320.9. 2-(4-Nitrophenyl)-N-4-methylbenzylacetamide. An N2purged flask equipped with a stirring bar was charged with 4-nitrophenylacetic acid (1.0 g, 5.52 mmol), 1,1′-carbonyldiimidazole (1.32 g, 8.1 mmol) and anhydrous CH2Cl2 (25 mL). The reddish-brown mixture was stirred for 45 min. 4-Methylbenzylamine (880 µL, 838 mg) was added via syringe. The cloudy yellow mixture was stirred for 1 h, then partitioned betwen CH2Cl2 and 1 N HCl. The organic layer containing a white precipitate was separated and washed with saturated aqueous NaHCO3. The precipitate was collected by suction filtration, triturated with MeOH and washed with ether to provide 0.78 g (53%) of the product as a white solid. MS (electrospray), m/e: 283.0 (M - H). 1H NMR (DMSO-d6): δ 2.30 (s, 3H), 3.68 (s, 2H), 4.26 (d, 2H, J ) 6.0 Hz), 7.15 (s, 4H), 7.58 (d, 1H, J ) 8.7 Hz), 8.22 (d, 1H, J ) 8.4 Hz), 8.65 (t, 1H, J ) 6.0 Hz). N-[2-(4-Nitrophenyl)acetyl]methanesulfonamide (50a). A suspension of 4-nitrophenylacetic acid (2.0 g, 11.0 mmol) in CH2Cl2 (30 mL) was treated with 1,1′-carbonyldiimidazole (CDI) (2.4 g, 11.0 mmol), yielding an orange solid. After the mixture was stirred for 1 h, methanesulfonamide (1.1 g, 11.0 mmol) was added and the mixture was stirred for an additional 1 h. Addition of diazobicyclo[5.4.0]undec-7-ene (1.5.5) (DBU) (1.7 mL, 11.0 mmol) yielded a purple mixture that was stirred for 16 h. The reaction mixture was then partitioned between 1 N aqueous HCl and EtOAc, during which time an orange solid fell out. The solid was partitioned between 1 N aqueous NaOH and EtOAc. The aqueous layer was acidified with 1 N aqueous HCl and extracted with EtOAc. The organic layer was dried over Na2SO4, filtered, and concentrated to yield 1.9 g of the product (67%) as a yellow solid, mp 191-193 °C. 1H NMR (DMSO): δ 3.21 (s, 3H), 3.79 (s, 2H), 7.52 (d, 2H, J ) 8.6 Hz), 8.17 (d, 2H, J ) 8.5 Hz). N-[2-(4-Nitrophenyl)acetyl]benzenesulfonamide (50b). The foregoing procedure was employed using benzenesulfon-

β3 Adrenergic Receptor Agonists amide (945 mg, 6.0 mmol), CDI (900 mg, 5.55 mmol), 4-nitrophenylacetic acid (1.0 g, 5.52 mmol), and DBU (0.87 mL, 5.81 mmol) in CH2Cl2 (20 mL) to yield 1.57 g (89%) of the product as a yellow solid, mp 145-148 °C. 1H NMR (400 MHz, DMSOd6): δ 3.73 (s, 2H), 7.40 (d, 2H, J ) 9 Hz), 7.69-7.53 (m, 3H), 7.87 (d, 2H, J ) 7 Hz), 8.11 (d, 2H, J ) 9 Hz), 12.45 (s, 1H). MS (ES) m/e: 319 (M - H). N-[2-(4-Nitrophenyl)acetyl]benzenesulfonamide (50c). The general procedure for 50a was employed using p-toluenesulfonamide (945 mg, 5.52 mmol), CDI (895 mg, 5.52 mmol), 4-nitrophenylacetic acid (1.0 g, 5.52 mmol), and DBU (0.84 g, 0.82 mL, 5.48 mmol) in CH2Cl2 (20 mL). The reaction yielded 1.74 g (94%) of the product as a yellow solid, mp 205-214 °C. Low-resolution 1H NMR (400 MHz, DMSO-d6): δ 2.35 (s, 3H), 3.71 (s, 2H), 7.41-7.36 (m, 4H), 7.75 (d, 2H, J ) 8 Hz), 8.11 (d, 2H, J ) 8 Hz), 12.35 (s, 1H). MS (ES) m/e: 33.0 (M - H). N-[2-(3-Nitrophenyl)acetyl]-4-methylbenzenesulfonamide (50d). The general procedure for 50a was employed using p-toluenesulfonamide (480 mg, 2.80 mmol), CDI (500 mg, 3.08 mmol), 3-nitrophenylacetic acid (450 mg, 2.48 mmol), and DBU (0.835 g, 0.82 mL, 5.48 mmol) in CH2Cl2 (15 mL). The product (600 mg, 65% yield) was obtained as a yellow solid, mp 175-177 °C. 1H NMR (400 MHz, DMSO- d6): δ 2.35 (s, 3H), 3.73 (s, 2H), 7.36 (d, 2H, J ) 8.0 Hz), 7.59-7.52 (m, 2H), 7.74 (d, 2H, J ) 8.0 Hz), 8.03 (s, 1H), 8.07 (dd, 1H, J ) 2.0, 8.0 Hz), 12.35 (s, 1H). MS (ES) m/e: 333.1 (M - H). 1-Nitro-3-[({[(phenylsulfonyl)amino]carbonyl}amino)methyl]benzene (56). Triethylamine (1.85 mL, 13.3 mmol) was added to 3-nitrobenzylamine (1.00 g, 5.30 mmol) in THF (26.5 mL), followed by benzenesulfonyl isocyanate (709.5 µL, 5.30 mmol), and the reaction mixture was stirred for 16 h at ambient temperature. Aqueous HCl (1 N) was added, and the mixture was extracted with EtOAc. The organic extracts were combined and concentrated. The residue was purified by silica gel chromatography, eluting with EtOAc/hexanes (4:1) to give 440 mg (25%) of the product. 1H NMR (300 MHz, CD3COCD3): δ 4.49 (d, 2H, J ) 7 Hz), 7.21 (br s, 1H), 7.53-7.68 (m, 5H), 7.99 (d, 2H, J ) 7 Hz), 8.09 (d, 1H, J ) 7 Hz), 8.10 (s, 1H), 9.73 (br s, 1H). MS (CI) m/e: 334 (M - H). Toluene-4-sulfonic Acid 2-(3-Nitrophenyl)ethyl Ester (51b).28 4-(Dimethylamino)pyridine (365.4 mg, 2.99 mmol) was added to 2-(3-nitrophenyl)ethanol (5.00 g, 29.91 mmol) in CHCl3 (60 mL) at ambient temperature, followed by triethylamine (5.84 mL, 38.7 mmol) and p-toluenesulfonyl chloride (6.84 g, 35.8 mmol). The reaction was stirred for 63 h, quenched with saturated Na2CO3, and extracted with CHCl3. The organic layer was dried and concentrated, and the residue was purified by silica gel chromatography, eluting with EtOAc/ hexanes (2:3) to give 6.14 g of the product (64%). 1H NMR (300 MHz, CDCl3): δ 2.40 (s, 3H), 3.04 (t, 2H, J ) 6 Hz), 4.26 (t, 2H, J ) 6 Hz), 7.24 (d, 2H, J ) 8 Hz), 7.42 (t, 1H, J ) 8 Hz), 7.47 (d, 1H, J ) 8 Hz), 7.62 (d, 2H, J ) 8 Hz), 7.87 (s, 1H), 8.05 (d, 1H, J ) 8 Hz). MS (CI) m/e: 344 (M + Na). Thioacetic Acid S-[2-(3-Nitrophenyl)ethyl] Ester (52a). Potassium thioacetate (4.36 g, 38.2 mmol) was added to tosylate 51b (6.14 g, 19.11 mmol) in acetonitrile (64 mL) at ambient temperature, and the reaction mixture was stirred for 17 h. The mixture was quenched with water and extracted with EtOAc, and the separated organic layer was dried, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with EtOAc/hexanes (1:4) to give 4.24 g of the product (98%). 1H NMR (300 MHz, CDCl3): δ 2.32 (s, 3H), 2.96 (t, 2H, J ) 7 Hz), 3.13 (t, 2H, J ) 7 Hz), 7.46 (t, 1H, J ) 8 Hz), 7.54 (d, 1H, J ) 7 Hz), 8.07 (s, 1H), 8.08 (d, 1H, J ) 7 Hz). MS (CI) m/e: 150 (M - AcS). 2-(3-Nitrophenyl)ethanesulfonic Acid (53a). Hydrogen peroxide (30% aqueous, 10.07 g, 5.0 equiv) was added to thioacetic acid S-[2-(3-nitrophenyl)ethyl]ester (52a, 4.24 g, 18.82 mmol) in acetic acid (38 mL) at ambient temperature, and the reaction mixture was stirred for 26 h. The mixture was concentrated, taken up in a small amount of toluene, and reconcentrated to give 4.22 g (97%) of the product. 1H NMR (300 MHz, CD3OD): δ 3.30 (dd, 2H, J ) 11, 7 Hz), 3.53 (dd,

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3 577 2H, J ) 11, 7 Hz), 7.63 (t, 1H, J ) 8 Hz), 7.82 (d, 1H, J ) 8 Hz), 8.12 (d, 1H, J ) 7), 8.23 (s, 1H). MS (CI) m/e: 230 (M H). 4-Nitrophenethylsulfonic Acid (53b). A suspension of 4-nitrophenethyl bromide (51a) (12.9 g, 56.1 mmol) and potassium thioacetate (11.4 g, 99.8 mmol) was stirred in DMSO (75 mL) for 24 h, diluted with EtOAc, extracted with water, dried, and concentrated to give crude 52b as a mobile brown oil (11.0 g). This material was dissolved in glacial acetic acid (75 mL), and 32% H2O2 (25 mL) was added. After 24 h, addition of water and concentration gave 10.8 g of a pale-yellow solid (84% crude yield) that was used without further purification. 1H NMR (300 MHz, DMSO-d6): δ 2.80 (t, 2H), 3.00 (t, 2H), 7.50 (d, 2H), 8.10 (d, 2H). 2-(3-Nitrophenyl)ethanesulfonic Acid Amide (54a). Thionyl chloride (19.7 mL, 20 equiv) was added to 2-(3nitrophenyl)ethanesulfonic acid (53a, 3.13 g, 13.53 mmol), and the resulting solution was heated at reflux for 17 h. The volatiles were removed under vacuum, NH3 in dioxane (135 mL, 0.5 M, 5.0 equiv) was added, and the reaction mixture was stirred for 5 h. Water was added followed by saturated aqueous Na2CO3, and the mixture was extracted with EtOAc. The extracts were dried over Na2SO4 and concentrated. The residue was purified by silica gel chromatography, eluting with EtOAc/hexanes (3:2) to give 1.83 g (59%) of the product. 1H NMR (300 MHz, CD3COCD3): δ 3.27-3.32 (m, 2H), 3.43-3.48 (m, 2H), 6.22 (s, 2H), 7.62 (t, 1H, J ) 8 Hz), 7.79 (d, 2H, J ) 8 Hz), 8.11 (d, 1H, J ) 8 Hz), 8.20 (d, 1H, J ) 2 Hz). MS (CI) m/e: 230 (M - H). 4-Nitrophenethylsulfonamide (54b). 4-Nitrophenethylsulfonic acid (53b, 23.1 g, 100 mmol) was heated at reflux with SOCl2 (200 mL) and N,N-DMF (10 mL). After 2 h the SOCl2 was removed under vacuum, and the residue was taken up in dioxane (200 mL) and added to 0.5 M NH3/dioxane (2000 mL). The mixture was filtered through Celite and then concentrated. The residue was dissolved in EtOAc (50 mL), and after addition of hexane, the product (17.07 g, 74%) precipitated as a tan solid. 1H NMR (300 MHz, DMSO-d6): δ 3.12 (t, 2H), 3.30 (t, 2H), 6.90 (s, 2H), 7.58 (d, 2H), 8.18 (d, 2H). MS (ES) m/e: 229 (M - H). 2-(3-Nitrophenyl)ethanesulfonic Acid Benzenesulfonylamide (55a). Triethylamine (508.7 µL, 3.64 mmol) was added to 2-(3-nitrophenyl)ethanesulfonic acid amide (54a, 420.2 mg, 1.83 mmol) in CH3CN (9 mL), followed by the addition of benzenesulfonyl chloride (279.4 µL, 2.19 mmol), and the reaction was heated at reflux for 4 h. Aqueous HCl (1 N) was then added, and the mixture was extracted with EtOAc. The combined organic layers were dried, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with MeOH/EtOAc (1:9) to give 362.5 mg (54%) of the product. 1H NMR (400 MHz, CD OD): δ 3.16-3.21 (m, 2H), 3.31-3.35 3 (m, 2H), 7.45-7.55 (m, 4H), 7.63 (d, 1H, J ) 8 Hz), 7.92-7.94 (m, 2H), 8.07-8.08 (m, 2H). MS (CI) m/e: 369 (M - H). 4-Nitrophenethyl-N-benzoylsulfonamide (55b). A mixture of 4-nitrophenethylsulfonamide (2.3 g, 10.0 mmol), CH3CN (40 mL), K2CO3 (2.80 g, 20.2 mmol), and benzoyl chloride (1.4 g, 10.0 mmol) was heated at reflux. After 3 h the mixture was diluted with water and EtOAc. Filtration gave the product (1.14 g) as a tan solid (34%). 1H NMR (300 MHz, DMSO-d6): δ 3.10 (t, 2H), 3.30 (m, 1H), 3.58 (t, 2H), 7.38 (t, 2H), 7.41 (m, 1H), 7.50 (d, 1H), 7.52 (d, 1H), 7.86 (d, 2H), 8.08 (d, 2H). MS m/e: 333 (M - H). 2-(4-Aminophenyl)-N-thiazol-2-ylacetamide (15). A slurry of 2-(4-nitrophenyl)-N-thiazol-2-ylacetamide (523 mg, 1.98 mmol) and 10% Pd/C (286 mg) in 40 mL of THF/ethanol (3:1) was stirred under 1 atm of H2 for 8 h. The mixture was filtered through a pad of Celite, which was washed with EtOAc. The filtrate was concentrated to afford 417 mg (90%) of the product as a yellow-orange solid. 1H NMR (CDCl3): δ 3.41 (s, 2H), 3.78 (bs, 2H), 6.39 (d, 1H), 6.66 (d, 1H), 6.86 (d, 1H), 7.14 (d, 1H), 11.22 (bs, 1H). MS (ES) m/e: 232 (M - H). N-[2-(4-Aminophenyl)acetyl]methanesulfonamide (16a). N-[2-(4-nitrophenyl)acetyl]methanesulfonamide (50a, 0.9 g, 3.5 mmol) was suspended in MeOH (25 mL) and treated with 10%

578

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3

Pd/C (750 mg) under positive N2 pressure. The reaction mixture was stirred under 1 atm of H2 for 7 h and then was filtered through a pad of Celite that was washed with MeOH. The filtrate was concentrated to give a crude product (750 mg, 94%), which was used without further purification. 1H NMR (DMSO-d6): δ 3.06 (s, 3H), 3.27 (s, 2H), 6.45 (d, 2H, J ) 8.2 Hz), 6.85 (d, 2H, J ) 8.2 Hz). N-[2-(4-Aminophenyl)acetyl]benzenesulfonamide (16b). The foregoing hydrogenation procedure was used with N-[2(4-nitrophenyl)acetyl]benzenesulfonamide (50b, 1.0 g, 3.12 mmol) and a reaction time of 3.5 h to give 470 mg of product (52%) as a yellow solid. 1H NMR (400 MHz, DMSO-d6): δ 3.25 (s, 2H), 3.28 (bs, 2H), 6.40 (d, 2H, J ) 8 Hz), 6.74 (d, 2H, J ) 8 Hz), 7.64-7.51 (m, 3H), 7.82 (d, 2H, J ) 8 Hz). MS (ES+) m/e: 313 (M + Na). N-2[2-(4-Aminophenyl)acetyl]benzenesulfonamide(16c). Tin(II) chloride hydrate (2.0 g, 8.86 mmol) was added to N-[2(4-nitrophenyl)acetyl]benzenesulfonamide (50c, 600 mg, 1.79 mmol) in 11.5:1 EtOH/N,N-DMF (33 mL), and the resulting mixture was heated at 70 °C for 1 h. The mixture was then poured onto ice, and the pH was adjusted to 8 with saturated aqueous NaHCO3. After addition of a small amount of EtOAc, the mixture was stirred at room temperature for 1 h. The layers were filtered through a pad of Celite and separated. The organic layer was dried, filtered, and concentrated to yield 140 mg of the product (26%) as a tan solid. 1H NMR (400 MHz, DMSO-d6): δ 2.31 (s, 3H), 3.14 (s, 2H), 6.75 (d, 2H, J ) 8 Hz), 7.25 (d, 2H, J ) 9 Hz), 7.64 (d, 2H, J ) 8 Hz), 7.92 (s, 3H). MS m/e: 305.1 (M + H). N-[2-(3-Aminophenyl)acetyl]-4-methylbenzenesulfonamide (17a). The foregoing procedure was used with (3nitrophenyl)acetyl]benzenesulfonamide (50d, 600 mg, 1.79 mmol) to give the product (230 mg, 42%). 1H NMR (400 MHz, DMSO-d6): δ 2.34 (s, 3H), 3.24 (s, 2H), 3.30 (2H + H2O), 5.00 (bs, 1H), 6.24 (d, 1H, J ) 8 Hz), 6.32 (s, 1H), 6.35 (d, 1H, J ) 8 Hz), 6.84 (t, 1H, J ) 8 Hz), 7.32 (d, 2H, J ) 8 Hz), 7.70 (d, 2H, J ) 8 Hz). MS (ES-) m/e: 303.1 (M - H). 1-Amino-3-[({[(phenylsulfonyl)amino]carbonyl}amino)methyl]benzene (17b). To 1-nitro-3-[({[(phenylsulfonyl)amino]carbonyl}amino)methyl]benzene (56, 431.1 mg, 1.29 mmol) was added 10% Pd/C (43.1 mg) in 3:1 MeOH/THF (12.8 mL) under an argon atmosphere. The mixture was stirred under 1 atm of H2 for 4 h at ambient temperature, then purged with N2, filtered through Celite, and concentrated to give 382.6 mg (98%) of the product. 1H NMR (300 MHz, CD3OD): δ 4.15 (s, 2H), 6.47 (d, 1H, J ) 8 Hz), 6.54 (s, 1H), 6.58 (d, 1H, J ) 8 Hz), 6.99 (t, 1H, J ) 8 Hz), 7.56 (t, 2H, J ) 7 Hz), 7.66 (t, 1H, J ) 7 Hz), 7.94 (d, 2H, J ) 7 Hz). MS (CI) m/e: 304 (M H). 2-(3-Aminophenyl)ethanesulfonic Acid Benzenesulfonylamide (17c). The foregoing procedure was used with 2-(3nitrophenyl)ethanesulfonic acid benzenesulfonylamide (55a, 362.5 mg, 978.7 µmol) and 10% Pd/C (36.2 mg) in MeOH (4.9 mL). Purification by silica gel chromatography, eluting with MeOH/EtOAc (1:9) gave 338.1 mg (99%) of product. 1H NMR (300 MHz, CD3OD): δ 2.90-3.01 (m, 2H), 3.22-3.26 (m, 2H), 6.50-6.57 (m, 2H), 6.67-6.81 (m, 1H), 6.97-7.12 (m, 1H), 7.43-7.51 (m, 3H), 7.92-7.94 (m, 2H). MS (CI) m/e: 339 (M H). 4-Aminophenethyl-N-benzoylsulfonamide (18). 4-Nitrophenethyl-N-benzoylsulfonamide (0.60 g) in MeOH (50 mL) and acetic acid (1 mL) was shaken on a Parr apparatus with 20% Pd(OH)2/C (0.2 g) under 45 psi of H2. When no further uptake was observed, the mixture was filtered through Celite and evaporated to give a white solid (0.60 g). 1H NMR (300 MHz, DMSO-d6): δ 2.90 (t, 2H), 3.70 (m, 2H), 6.61 (d, 2H), 6.98 (d, 2H), 7.42 (m, 2H), 7.60 (m, 1H), 7.80 (m, 2H). MS (ES) m/e: 303 (M - H). 2-(4-Aminophenyl)-N-4-methylbenzylacetamide (19a). An N2-purged flask was charged with absolute EtOH (40 mL), 2-(4-nitrophenyl)-N-4-methylbenzylacetamide (515 mg, 1.81 mmol), and 10% Pd/C (312 mg). The mixture was placed under 1 atm of H2 and stirred for 16 h, then filtered through a pad of Celite, washing with additional ethanol. The filtrate was

Uehling et al. concentrated to supply the crude product as an off-white solid. Purification by silica gel chromatography (eluting with 1:1 hexane/EtOAc) afforded 303 mg (66% yield) of the product as a white solid. 1H NMR (CDCl3): δ 2.27 (s, 3H), 3.48 (s, 2H), 3.62 (bs, 2H), 4.32 (d, 2H, J ) 6.0 Hz), 5.60 (bs, 1H), 6.61 (d, 2H, J ) 8.4 Hz), 6.99 (d, 2H, J ) 8.4 Hz), 7.02 (d, 2H, J ) 8.8 Hz), 7.06 (d, 2H, J ) 8.0 Hz). MS (ES) m/e: 255.1 (M + H). N-[2-(4-Aminophenyl)ethyl]-4-methylbenzenesulfonamide (19b).26 The foregoing procedure was employed with N-[2-(4-nitrophenyl)ethyl]-4-methylbenzenesulfonamide (497 mg, 1.55 mmol) and 10% Pd/C (299 mg) in absolute EtOH (35 mL). Crude product was purified by silica gel chromatography (eluting with 3:2 hexane/EtOAc) to afford 181 mg (40% yield) of the product as a pink solid. 1H NMR (CDCl3): δ 2.39 (s, 3H), 2.60 (t, 2H, J ) 6.8 Hz), 3.10 (q, 2H, J ) 6.8 Hz), 4.21 (m, 1H), 6.55 (d, 2H, J ) 8.0 Hz), 6.81 (d, 2H, J ) 8.0 Hz), 7.25 (d, 2H, J ) 8.0 Hz), 7.64 (d, 2H, J ) 8.0 Hz). General Procedure for Synthesis of Final Targets via Reductive Amination of Anilines with tert-Butyl (2R)2-{[tert-Butyl(dimethyl)silyl]oxy}-2-(3-chlorophenyl)ethyl[(1R)-1-methyl-2-oxoethyl]carbamate (10) and Deprotection. Procedure A. A solution of aldehyde 10 in CH2Cl2 (0.10.2 M) and aniline intermediate (0.67-1.2 equiv) was stirred for 10-30 min, and NaBH(OAc)3 (2.0-2.5 equiv), followed by catalytic glacial acetic acid (1-10 drops), was added. The resulting mixture was stirred for a period of 8-48 h, then added to CH2Cl2 and saturated aqueous Na2CO3. The organic layer was separated, and the aqueous layer was extracted with CH2Cl2. The combined organic layers were dried over Na2SO4, filtered, and concentrated to give the crude aniline intermediate, which was purified by silica gel chromatography. The resulting aniline was subjected to acidic deprotection with 4 N HCl/dioxane or 6 N aqueous HCl. The crude target hydrochloride, which was obtained by concentration and/or precipitation with Et2O, was purified by silica gel chromatography using a mixture of CHCl3, MeOH, and concentrated NH4OH to supply the target as the free base. Procedure B. A procedure similar to procedure A was followed except that the aniline and aldehyde 10 were dissolved in MeOH and an excess of NaCN(BH)3 and catalytic acetic acid were added. The mixture was stirred for 12-86 h and worked up by partitioning between water and EtOAc. The organic layer was separated and dried to give the crude intermediate that was deprotected as described in the foregoing procedure. Methyl 4-[((2R)-2-{(tert-Butoxycarbonyl)[(2R)-2-{[tertbutyl(dimethyl)silyl]oxy}-2-(3-chlorophenyl)ethyl]amino}propyl)amino]benzoate (Intermediate 20). Procedure B was employed using methyl 4-aminobenzoate (430 mg, 2.84 mmol), aldehyde 10 (1.5 g, 3.41 mmol), acetic acid (650 mg, 10.8 mmol), MeOH (25 mL), and NaCNBH3 (0.45 g, 7.41 mmol). The reaction mixture was stirred for 3 days, the reaction was quenched with 15% w/v solution of potassium sodium tartrate, the mixture was then stirred for 30 min, and the reaction was worked up. The crude product was purified by chromatography, eluting with 9:1 hexane/EtOAc, to afford 932 mg of intermediate 20 (59%) as a foamy oil. 1H NMR (CDCl3): δ -0.17 (s, 3H), -04 to 0.02 (m, 3H), 0.77-0.86 (m, 12H), 1.40 (s, 9H), 3.03-3.25 (m, 4H), 3.78 (s, 3H), 4.03-4.07 (m, 1H), 6.45 (d, 2H, J ) 8.6 Hz), 7.09-7.26 (m, 4H), 7.79 (d, 2H, J ) 8.6 Hz). MS (ES) m/e: 577 (M + H)+. 4-[((2R)-2{[(2R)-2-(3-Chlorophenyl)-2-hydroxyethyl]amino}propyl)amino]benzoic Acid (36). Intermediate 20 (466 mg, 0.81 mmol) was treated with 4 N HCl/dioxane (10 mL) for 16 h to give, after addition of Et2O, 323 mg (85%) of methyl 4-[((2R)2-{[(2R)-2-(3-chlorophenyl)-2-hydroxyethyl]amino}propyl)amino]benzoate hydrochloride as a white solid. 1H NMR (CD3OD): δ 1.37 (d, 3H, J ) 6.4 Hz), 3.10-3.73 (m, 6H), 3.78 (s, 3H), 4.98 (dd, 1H, J ) 2.9, 10 Hz), 6.71 (d, 2H, J ) 8.8 Hz), 7.27 (m, 3H), 7.44 (s, 1H), 7.79 (d, 2H, J ) 8.8 Hz). MS (ES) m/e: 363 (M + H). A portion (253 mg, 0.63 mmol) of the methyl ester obtained above was dissolved in MeOH/water (12 mL) and treated with LiOH‚H2O (146 mg, 3.48 mmol) in water (4 mL). The mixture

β3 Adrenergic Receptor Agonists was stirred for 16 h at ambient temperature, heated to 40 °C for 7 h, then heated at 60 °C for 16 h. The mixture was allowed to cool to ambient temperature, and the solvent was removed. The residue was purified by chromatography eluting with CHCl3/MeOH/concentrated NH4OH (6:1:0.1) to afford 172 mg of acid 36 (78%) as a white solid judged by 1H NMR analysis to be a 5.5:1 mixture of R,R/minor diastereomers. 1H NMR (400 MHz, CD3OD) (R,R diastereomer): δ 1.26 (d, 3H, J ) 6.9 Hz), 3.01-3.09 (m, 2H), 3.30-3.38 (m, 3H), 6.62 (d, 2H, J ) 8.6 Hz), 7.24-7.39 (m, 3H), 7.41 (s, 1H), 7.76 (d, 2H, J ) 8.6 Hz). HRMS ((M + H)+ found: 349.1300. Calcd for C18H21Cl1N2O3: 349.1319. Anal. (C18H21Cl1N2O3‚1.5H2O) C, H, N. tert-Butyl (1R)-2-Anilino-1-methylethyl-[(2R)-2-{[tertbutyl(dimethyl)silyl]oxy}-2-(3-chlorophenyl)ethyl]carbamate (Intermediate 21). Procedure A was followed using aldehyde 10 (270.6 mg, 0.61 mmol), aniline (65 µL (0.713 mmol), NaBH(OAc)3 (305 mg, 1.44 mmol), and 1 drop of AcOH in CH2Cl2 (4.0 mL). Workup gave the crude product, which was purified by chromatography (eluting with 10:1 followed by 5:1 hexane/ethyl acetate) to give 274.0 mg (74%) of the product as a colorless oil. 1H NMR (400, CDCl3): δ -0.12 (s, 3H), 0.03 (s, 3H), 0.87 (m, 12H), 1.45 (s, 9H), 3.11-3.20 (m, 4H), 6.55 (d, 2H, J ) 10.8 Hz), 6.67 (t, 1H, J ) 10.8), 7.137.26 (m, 5H), 7.32 (s, 1H). (1R)-2-{[(1R)-2-Anilino-1-methylethyl]amino}-1-(3-chlorophenyl)ethanol Hydrochloride (39). The product of the foregoing procedure was dissolved in CH2Cl2 (5 mL), and TFA (1 mL) was added. The resulting mixture was stirred for 45 min and concentrated. The residue was taken up in 1:1 THF/6 N aqueous HCl (10 mL), and the mixture was stirred for 16 h. Concentration followed by chromatography (eluting with 214: 15:1 CHCl3/MeOH/concentrated NH4OH) gave 119.1 mg of the free base product. This material was dissolved in 1:1 CH3CN/ H2O (30 mL), and 2 mL of 1.0 N HCl was added. The mixture was lyophilized to afford 136.9 mg (76% yield) of the product hydrochloride judged by 1H NMR analysis to be a 9.8:1 mixture of R,R/minor diastereomers. 1H NMR (400 MHz, CD3OD) (R,R diastereomer): δ 1.47 (d, 3H, J ) 6.4 Hz), 3.20 (dd, 1H, J ) 13.8, 6.0 Hz), 3.53 (dd, 1H, J ) 13.8, 6.0 Hz), 3.70 (s, 1H), 5.04 (dd, 1H, J ) 10.0, 2.8 Hz), 7.05 (t, 1H, J ) 7.6 Hz), 7.347.38 (m, 5H), 7.50 (s, 1H, 3H), 6.62 (d, 2H, J ) 8.6 Hz), 7.247.39 (m, 3H), 7.41 (s, 1H), 7.76 (d, 2H, J ) 8.6 Hz). MS (ES) m/e: 305.3 (M + H). Anal. (C17H21Cl1N2O‚2.0HCl‚0.5H2O) C, H, N. tert-Butyl (1R)-2-{4-[(Acetylamino)methyl]anilino}-1methylethyl-[(2R)-2-{[tert-butyl(dimethyl)silyl]oxy}-2-(3chlorophenyl)ethyl]carbamate (Intermediate 22). General procedure A was followed using aldehyde 10 (1.68 g, 3.80 mmol), N-(4-aminobenzyl)acetamide (13, 500 mg, 3.04 mmol), 2 drops of glacial acetic acid, and NaBH(OAc)3 (1.52 g, 7.17 mmol). The reaction time was 8 h. Workup and chromatography (eluting with 1:1 hexane/EtOAc) afforded 1.18 g (67%) of the intermediate 22. 1H NMR (CDCl3): δ -0.16 (s, 3H), -0.03 (s, 3H), 0.82 (m, 12H), 1.41 (s, 9H), 1.95 (s, 3H), 3.03-3.19 (m, 4H), 4.05-4.11 (m, 1H), 4.25 (d, 2H, J ) 5.6 Hz), 4.93 (bs, 0.5H), 5.20 (bs, 0.5H), 6.47 (d, 2H, J ) 8.8 Hz), 7.03 (d, 2H, J ) 8.4 Hz), 7.18-7.22 (m, 3H), 7.27 (s, 1H). MS (ES) m/e: 590.3, 591.6 (M + H). N-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]benzyl}acetamide (37). Intermediate 22 (580 mg, 0.982 mmol) was treated with 4 N aqueous HCl/dioxane (10 mL) for 3 h and worked up according to the general procedure to supply an amorphous material, which was dissolved in a small amount of 30:15:1 CHCl3/MeOH/ concentrated NH4OH and purified by chromatography (eluting with 5:1 EtOAc/MeOH) to afford 256 mg of product 37 (69%) as a colorless glass judged by 1H NMR analysis to be a 6:1 mixture of R,R/minor diastereomers. 1H NMR (CD3OD) (major diastereomer): δ 1.27 (d, 3H, J ) 6.8 Hz), 1.91 (s, 3H), 3.023.08 (m, 2H), 3.22-3.33 (m, 1H), 4.17 (s, 2H), 6.62 (d, 2H, J ) 8.4 Hz), 7.04 (d, 2H, J ) 8.4 Hz), 7.27-7.38 (m, 3H), 7.40 (s, 1H). HRMS (M + H)+ found: 376.1768. Calcd for C20H26Cl1N3O2: 376.1792. Anal. (C20H26Cl1N3O2‚1H2O‚0.5CHCl3) C, H, N.

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3 579 tert-Butyl (2R)-2-{[tert-butyl(dimethyl)silyl]oxy}-2-(3chlorophenyl)ethyl[(1R)-1-methyl-2-(4-{[(methylsulfonyl)amino]methyl}anilino)ethyl]carbamate (Intermediate 23). General procedure A was followed using aldehyde 10 (1.68 g, 3.80 mmol), N-(4-aminobenzyl)methanesulfonamide hydrochloride (14, 720 mg, 3.04 mmol), acetic acid (2 drops), and NaBH(OAc)3 (1.50 g, 7.08 mmol). Workup and purification by chromatography (eluting with 2:1 hexane/EtOAc) afforded 1.32 g (69%) of intermediate 23 as a colorless glass. 1H NMR (CDCl3): δ -0.16 (s, 3H), -0.01 (s, 3H), 0.83 (m, 12H), 1.53 (s, 9H), 2.66 (s, 1.5H), 2.67 (s, 1.5H), 2.95-3.20 (m, 4H), 3.804.00 (m, 2H), 4.09 (s, 1H), 4.10 (s, 1H), 6.49 (d, 2H, J ) 8.4 Hz), 7.11 (d, 2H, J ) 8.4 Hz), 7.18-7.22 (m, 4H). MS (ES) m/e: 626.2, 628.4 (M + H). N-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]benzyl}methanesulfonamide (38). Intermediate 23 (410 mg, 0.655 mmol) was stirred in 4 N HCl/ dioxane (10 mL) for 3 h and worked up according to the general procedure to supply the crude material, which was dissolved in a small amount of CHCl3/MeOH/concentrated NH4OH (30: 15:1) and purified by chromatography (eluting with 5:1 EtOAc/ MeOH) to supply 239 mg (89%) of target 38 as a colorless glass that was judged by 1H NMR to be a 10:1 mixture of R,R/minor diastereomers. 1H NMR (MeOD-d4): δ 1.20 (d, 3H, J ) 7.2 Hz), 2.57 (s, 3H), 2.95-2.97 (m, 2H), 3.18-3.20 (m, 1H) 4.13 (s, 2H), 4.78-4.83 (m, 1H), 6.62 (d, 2H, J ) 8.4 Hz), 7.14 (d, 2H, J ) 8.4 Hz), 7.25-7.29 (m, 3H), 7.39 (s, 1H). HRMS (M + H)+ found: 412.1455. Calcd for C19H26Cl1N3O3S1: 412.1462. Anal. (C19H26Cl1N3O3S1‚0.5H2O‚0.25CHCl3) C, H, N. tert-Butyl (2R)-2-{[tert-Butyl(dimethyl)silyl]oxy}-2-(3chlorophenyl)ethyl-((1R)-1-methyl-2-{4-[2-oxo-2-(1,3-thiazol-2-ylamino)ethyl]anilino}ethyl)carbamate (Intermediate 24). General procedure A was employed using aldehyde 10 (212 mg, 0.482 mmol), 2-(4-aminophenyl)-N-thiazol-2ylacetamide (15, 215 mg, 0.922 mmol), acetic acid (2 drops), and NaBH(OAc)3 (230 mg, 1.09 mmol) in 3:1 CH2Cl2/N,N-DMF (8 mL). The intermediate 24 (267 mg, 82%) was obtained as a pale-yellow oil following workup and chromatographic purification (eluting with 2:1 to 1:1 hexane/EtOAc). 1H NMR (CDCl3): δ -1.15 (s, 3H), -0.01 (s, 3H), 0.80 (m, 12H), 1.43 (s, 9H), 3.00-3.21 (m, 4H), 3.80-4.00 (m, 2H), 3.62 (s, 2H), 3.80-4.00 (m, 2H), 6.50 (d, 2H, J ) 8 Hz), 6.95 (d, 1H, J ) 2 Hz), 7.03 (d, 2H, J ) 8 Hz), 7.10-7.35 (m, 4H), 7.39 (s, 1H). MS (ES) m/e: 657 (M - H). 2-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}-N-(1,3-thiazol-2-yl)acetamide (40). To a solution of intermediate 24 (267 mg, 0.405 mmol) in THF (10 mL) was added 1.0 M tetrabutylammonium fluoride/THF (610 µL, 0.610 mmol), and the mixture was stirred for 16 h. The mixture was partitioned between EtOAc and water, and the organic layer was separated, dried over Na2SO4, filtered, and concentrated to provide 145 mg of a residue that was dissolved in CH2Cl2 (5 mL) and TFA (0.5 mL). The mixture was stirred for 16 h and concentrated. The residue was purified by chromatography (eluting with 150:30:1 CHCl3/ MeOH/concentrated NH4OH) to afford 93.9 mg (52%) of the product as a bright-yellow solid judged by 1H NMR analysis to be a 11:1 mixture of R,R/minor diastereomers. 1H NMR (CDCl3) (R,R diastereomer): δ 1.12 (d, 3H, J ) 6.0 Hz), 2.74 (dd, 1H, J ) 9.2, 12.2 Hz), 2.89 (dd, 1H, J ) 3.6, 12.2 Hz), 2.93-3.01 (m, 2H), 3.15 (d, 1H, J ) 8.4 Hz), 3.70 (s, 2H), 4.11 (bs, 1H), 4.63 (dd, 1H, J ) 3.6, 8.8 Hz), 6.59 (d, 2H, J ) 8.4 Hz), 6.92 (d, 1H, J ) 3.6 Hz), 7.07 (d, 2H, J ) 8.4 Hz), 7.187.26 (m, 3H), 7.32 (d, 1H, J ) 3.6 Hz), 7.35 (s, 1H). HRMS (M + H)+ found: 445.1485. Calcd for C22H25Cl1N4O2S1: 445.1465. Anal. (C22H25Cl1N4O2S1) C, H, N. tert-Butyl (2R)-2-{[tert-Butyl(dimethyl)silyl]oxy}-2(3-chlorophenyl)ethyl[(1R)-1-methyl-2-(4-{2-[(methylsulfonyl)amino]-2-oxoethyl}anilino)ethyl]carbamate (Intermediate 25). General procedure A was employed using aldehyde 10 (720 mg, 1.6 mmol), N-[2-(4-aminophenyl)acetyl]methanesulfonamide (16a, 750 mg, 3.3 mmol), and NaBH(OAc)3 (1.0 g, 4.7 mmol) in CH2Cl2/N,N-DMF (8:3). After the mixture was stirred for 2.5 days, 15% w/v aqueous potassium

580

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3

sodium tartrate was added and standard workup gave intermediate 25 (1.0 g, 95%). 1H NMR (DMSO-d6): δ -0.16 (s, 3H), -0.19 (s, 3H), 0.70-0.89 (m, 12H), 1.28-1.32 (m, 9H), 2.04 (2, 2H), 2.46-2.47 (m, 5H), 6.39-6.45 (m, 1H), 6.91 (d, 1H, J ) 8.4 Hz), 7.17-7.34 (m, 5H), 7.92 (s, 1H). N-(2-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}acetyl)methanesulfonamide (41). Intermediate 25 (1.01 g, 1.5 mmol) was dissolved in 4 N HCl/dioxane (10 mL), and the mixture was stirred for 3 h. The solvent was removed, and the residue was purified by chromatography, eluting with CHCl3/MeOH/ concentrated NH4OH (3:1:0.1 to 3:1.5:0.1), to afford target 41 as a clear oil (380 mg, 56%) judged by 1H NMR to be a 12.3:1 mixture of R,R/minor diastereomers. 1H NMR (DMSO-d6) (R,R diastereomer): δ 1.08 (d, 3H, J ) 5.9 Hz), 2.79-3.09 (m, 4 H), 3.13 (s, 3H), 3.17 (s, 2H), 4.07 (broad s, 1H), 4.71-4.77 (m, 1H), 6.48 (d, 2H, J ) 8.2 Hz), 6.92 (d, 2H, J ) 8.2 Hz), 7.30-7.36 (m, 3H), 7.40 (s, 1H). MS(ES) m/e: 440 (M + H)+, 462 (M + Na)+. HRMS (M + H)+ found: 440.1407. Calcd for C20H26N3O4S1Cl1: 440.1411. Anal. (C20H26N3O4S1Cl1‚ 1.25H2O): C, H, N. N-(2-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}acetyl)benzenesulfonamide (42). General procedure A was employed using aldehyde 10 (187 mg, 0.423 mmol), N-[2-(4-aminophenyl)acetyl]benzenesulfonamide (16b, 187 mg, 0.644 mmol), NaBH(OAc)3 (850 mg, 4.01 mmol), and acetic acid (3 drops) in 1:2 CH2Cl2/N,N-DMF (30 mL). After 16 h, the reaction was quenched with aqueous 15% w/v potassium sodium tartrate and worked up as described in the general procedure to afford, after chromatography (eluting with 7:3 hexane/EtOAc), the Boc amine silyl ether intermediate 26. This material was dissolved in 1:1 6 N aqueous HCl/THF (6 mL), and the resulting solution was stirred for 5 h. Removal of volatiles afforded a residue that was purified by chromatography, eluting with CHCl3/ concentrated MeOH/NH4OH (6:1:0.1), to supply the product (7 mg, 18%) as an off-white solid judged by 1H NMR analysis to be a 26:1 mixture of R,R/minor diastereomers. 1H NMR (400 MHz, CD3OD) (R,R diastereomer): δ 1.33 (d, 3H, J ) 6.4 Hz), 3.06-3.19 (m, 3H), 3.24-3.31 (m, 3H), 4.92 (dd, 1H, J ) 9.8, 2.8 Hz), 6.57 (d, 2H, J ) 8.4 Hz), 7.02 (d, 2H, J ) 8.4), 7.317.49 (m, 7H), 7.88 (d, 2H, J ) 7.2). HRMS (M + H)+ found: 502.1532. Calcd for C25H28N3O4S1Cl1: 502.1567. Anal. (C25H28N3O4S1Cl1‚1.25H2O): C, H, N. N-({4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}acetyl)-4-methylbenzenesulfonamide (43). General procedure A was employed using aldehyde 10 (169 mg, 0.382 mmol), N-[2-(4-aminophenyl)acetyl]-4-methylbenzenesulfonamide (16c, 140 mg, 0.460 mmol), NaBH(OAc)3 (602 mg, 2.84 mmol), and acetic acid (3 drops) in CH2Cl2 (22 mL). Workup gave a residue that was purified by chromatography, eluting with hexane/EtOAc (7:3) to provide intermediate 27 as white foam (152 mg). This material was dissolved in 4 N aqueous HCl (10 mL), and the resulting solution was stirred for 6 h to give, after addition of Et2O, an off-white solid. This material was dissolved in CHCl3/MeOH/ concentrated NH4OH (6:1:0.2) and purified by chromatography (eluting with 6:1:0.1 CHCl3/MeOH/concentrated NH4OH) to yield 35 mg (33%) of product 43 as an off-white solid, mp 100105 °C, judged by 1H NMR to be >95% R,R diastereomer. 1H NMR (400 MHz, CD3OD): δ 1.36 (d, 3H, J ) 6.4 Hz), 2.38 (s, 3H), 3.12-3.14 (m, 1H), 3.20 (dd, 1H, J ) 12.6, 2.8 Hz), 3.303.40 (m, 1H), 3.49 (m, 1H), 4.94 (dd, 1H, J ) 10.0, 3.2 Hz), 6.60 (d, 2H, J ) 8.0 Hz), 7.01 (d, 2H, J ) 8.4 Hz), 7.27 (d, 2H, J ) 8.0 Hz), 7.32-7.35 (m, 3H), 7.47 (s, 1H), 7.78 (d, 2H, J ) 8.4 Hz). HRMS (M + H)+ found: 516.1734. Calcd for C26H30N3O4S1Cl1: 516.1724. Anal. (C26H30N3O4S1Cl1‚ 0.25CHCl3): C, H, N. N-({3-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}acetyl)-4-methylbenzenesulfonamide (44). General procedure A was employed using aldehyde 10 (217 mg, 0.491 mmol), N-[2-(3-aminophenyl)acetyl]benzenesulfonamide (17a, 180 mg, 0.591 mmol), NaBH(OAc)3 (207 mg, 0.977 mmol), and acetic acid (3 drops) in

Uehling et al. CH2Cl2 (22 mL). After 16 h the reaction was worked up and the crude material was purified by chromatography, eluting with hexane/EtOAc (7:3 to 1:1) to yield intermediate 28 as a white foam (112 mg, 31%). The foam was dissolved in 4 N HCl/ dioxane (10 mL), and the resulting solution was stirred for 16 h to give, after workup, a sticky solid. Purification by chromatography (eluting with 6:1:0.1 CHCl3/MeOH/concentrated NH4OH) gave 31 mg (40% from 10) of the target 44 as an offwhite solid, mp 105-110 °C, judged by 1H NMR to be >95% R,R diastereomer. 1H NMR (400 MHz, DMSO-d6): δ 1.26 (d, 3H, J ) 7 Hz), 2.32 (s, 3H), 2.98-3.03 (m, 1H), 3.11-3.18 (m, 2H), 3.29 (s, 2H), 3.41-3.46 (m, 1H), 4.94 (dd, 1H, J ) 10, 3 Hz), 6.48-6.52 (m, 2H), 6.59 (s, 1H), 6.96 (t, 1H, J ) 8 Hz), 7.18 (d, 2H, J ) 8 Hz), 7.27-7.30 (m, 3H), 7.42 (s, 1H), 7.73 (d, 2H, J ) 8 Hz). HRMS (M + H)+ found: 516.1728. Calcd for C26H30N3O4S1Cl1: 516.1724. Anal. (C26H30N3O4S1Cl1‚ 0.1CHCl3‚0.85CH3OH): C, H, N. 1-[((2R)-2-{[(2R)-2-(3-Chlorophenyl)-2-hydroxyethyl]amino}propyl)amino]-3-[({[(phenylsulfonyl)amino]carbonyl}amino)methyl]benzene (45). General procedure B was employed using aldehyde 10 (461.6 mg, 1.04 mmol), and 1-amino-3-[({[(phenylsulfonyl)amino]carbonyl}amino)methyl]benzene (17b, 382.6 mg, 1.25 mmol) in MeOH (10.4 mL). The mixture was heated to 50 °C, and NaCNBH3 (44.0 mg, 0.70 mmol) and acetic acid (45.0 µL, 0.79 mmol) were added. After the mixture was stirred for 17 h, the reaction was quenched by addition of a 15% aqueous w/v solution of potassium sodium tartrate and worked up to yield the crude material. Purification by chromatography, eluting with EtOAc/hexanes (2:3), supplied intermediate 29. This material was dissolved in 4 N HCl/dioxane (7.8 mL), and the mixture was stirred for 3 h. Workup gave a material that was purified by chromatography, eluting with CHCl3/MeOH (4:1), to afford 184.2 mg (34%) of target 45 as a tan solid judged by 1H NMR analysis to be a 7.7:1 mixture of R,R/minor diastereomers. 1H NMR (300 MHz, DMSO-d6) (R,R diastereomer): δ 1.11 (d, 3H, J ) 5.6), 2.863.40 (m, 5H), 4.00 (bs, 2H), 4.77 (m, 1H), 5.58 (bs, 1H), 6.35 (d, 1H, J ) 7.2 Hz), 6.42 (d, 2H, J ) 8.0 Hz), 6.59 (bs, 1H), 6.95 (t, 1H, J ) 7.6 Hz), 7.33-7.38 (m, 4H), 7.43-7.48 (m, 3H), 7.80 (d, 2H, J ) 6.8 Hz). HRMS (M + H)+ found: 517.1696. Calcd for C25H29N4O4S1Cl1: 517.1676. Anal. (C25H29N4O4S1Cl1‚ 0.75H2O): C, H, N. 3-{4-[((2R)-2-{[(2R)-2-(3-Chlorophenyl)-2-hydroxyethyl]amino}propyl)amino]phenyl}propanoic Acid (35). General procedure A was employed using aldehyde 10 (520 mg, 1.18 mmol), 3-(4-aminophenyl)propanoic acid (233 mg, 1.41 mmol), NaBH(OAc)3 (394 mg, 1.76 mmol), glacial acetic acid (81.0 µL, 1.41 mmol), and 1,2-dichloroethane as solvent. The reaction mixture was stirred for 27.5 h and worked up to give intermediate 30 (332 mg). A portion of this material (321 mg) was dissolved in 4 N HCl/dioxane (4.0 mL). After 5 h, the solution was concentrated and neutralized by addition of 3 drops of concentrated NH4OH. The residue was purified by chromatography, eluting with CHCl3/MeOH/concentrated NH4OH (80:15:2, then 60:15:2), to afford material that was dissolved in EtOH/H2O (1:1). Lyophilization gave 137 mg (31% overall) of target 35 judged by 1H NMR to be a 5.5:1 mixture of R,R/minor diasteromers. 1H NMR (300 MHz, DMSO-d6) (R,R diastereomer): δ 1.04 (d, 3H, J ) 5.8 Hz), 2.40-2.46 (m, 2H), 2.64-2.69 (m, 2H), 2.72 (d, 2H, J ) 6.2 Hz), 2.80-2.90 (m, 3H), 3.40 (bs, 2H), 4.64 (m, 1H), 5.30 (bs, 1H), 6.49 (d, 2H, J ) 8.4 Hz), 6.92 (d, 2H, J ) 8.4 Hz), 7.29-7.39 (m, 3H), 7.42 (s, 1H). Anal. (C20H25N2O3Cl1‚0.75H2O‚0.1EtOH): C, H, N. 2-{3-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}-N-(phenylsulfonyl)ethanesulfonamide (46). General procedure B was employed using aldehyde 10 (351.2 mg, 794.6 µmol), (2-(3-aminophenyl)ethanesulfonic acid benzenesulfonylamide (17c, 338.1 mg, 0.993 mmol), NaCNBH3 (41.8 mg, 0.67 mmol), and acetic acid (42.8 µL, 0.75 mmol). The reaction mixture was stirred for 86 h, quenched with 15% w/v solution of potassium sodium tartrate, and worked up to supply intermediate 31, which was purified by chromatography, eluting with MeOH/EtOAc (1:4). This material was dissolved in 4 N HCl in dioxane (5.5 mL),

β3 Adrenergic Receptor Agonists and the solution was stirred for 4 h. After concentration, the residue was purified by chromatography, eluting with CHCl3/ MeOH (3:1) containing concentrated NH4OH, to give 142.7 mg (33%) of product 46 judged by 1H NMR to be >95% R,R diastereomer. 1H NMR (400 MHz, CD3OD): δ 1.38 (d, 3H, J ) 6.8 Hz), 2.94-3.00 (m, 2H), 3.11-3.17 (m, 1H), 3.21-3.37 (m, 3H), 3.48-3.55 (m, 3H), 4.96 (dd, 1H, J ) 10.2, 2.8 Hz), 6.65 (d, 2H, J ) 8.4 Hz), 7.04 (dd, 2H, J ) 8.4 Hz), 7.30-7.46 (m, 7H), 8.00 (d, 2H, J ) 7.2 Hz). HRMS (M + H)+ found: 552.1422. Calcd for C25H31Cl1N3O5S2: 552.1394. Anal. (C25H31Cl1N3O5S2‚1.0H2O): C, H, N. N-Benzoyl-2-{4-[((2R)-2-{[(2R)-2-(3-chlorophenyl)-2hydroxyethyl]amino}propyl)amino]phenyl}ethanesulfonamide (47). General procedure B was employed using aldehyde 10 (1.05 g, 2.38 mmol), 4-aminophenethyl-N-benzoylsulfonamide (18, 1.2 g, 3.94 mmol), glacial acetic acid (0.3 mL), and NaCNBH3 (250 mg) in MeOH (25 mL). Workup afforded crude protected intermediate 32, which was stirred with 4 N HCl/dioxane for 4 h. The solvent was evaporated, and the residue was treated with aqueous Na2CO3 and EtOAc. The mixture was then filtered to give product 47 as a white solid (110 mg, 9% yield), which was judged by 1H NMR to be a 22:1 mixture of R,R/minor diastereomers. 1H NMR (400 MHz, CD3OD) (R,R diastereomer): δ 1.11 (d, 3H, J ) 5.6 Hz), 2.85-3.39 (m, 5H), 4.00 (bs, 1H), 4.68-4.88 (m, 1H), 5.55 (bs, 1H), 6.35 (d, 1H, J ) 7.2 Hz), 6.58 (bs, 1H), 6.95 (t, 1H, J ) 7.6 Hz), 7.32-7.38 (m, 4H), 7.43-7.48 (m, 3H), 7.80 (d, 2H, J ) 6.8 Hz). MS m/e: 515 (M - H). Anal. (C26H30N3O4S1Cl1‚ 1.0H2O): C, H, N. tert-Butyl (2R)-2-{[tert-Butyl(dimethyl)silyl]oxy}-2-(3chlorophenyl)ethyl[(1R)-1-methyl-2-(4-{2-[(4-methylbenzyl)amino]-2-oxoethyl}anilino)ethyl]carbamate (Intermediate 33). General procedure A was employed using aldehyde 10 (459 mg, 1.30 mmol), 2-(4-aminophenyl)-N-4methylbenzylacetamide (19a, 264 mg, 1.04 mmol), acetic acid (3 drops), and NaBH(OAc)3 (482 mg, 2.28 mmol) in CH2Cl2 (10 mL). The mixture was stirred for 16 h and worked up. Chromatography (eluting with 5:1 hexanes/EtOAc) supplied 583 mg (82%) of intermediate 33. 1H NMR (CDCl3): δ -0.09 (s, 3H), 0.06 (s, 3H), 0.90 (s, 12H), 1.48 (s, 9H), 2.35 (s, 3H), 3.10-3.30 (m, 4H), 3.55 (S, 3H), 4.1 (bs, 1H), 4.39 (d, 2H, J ) 5.7 Hz), 6.55 (d, 2H, J ) 8.4 Hz), 7.06 (d, 2H, J ) 8.4 Hz), 7.10-7.40 (m, 7H). MS (ES), m/e: 679.9, 681.9 (M + H). 2-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}-N-(4-methylbenzyl)acetamide (48). Intermediate 33 (583 mg, 0.857 mmol) was dissolved in 4 N HCl/dioxane (10 mL), and the reaction mixture was stirred for 3 h. Approximately 50 mL of Et2O was added, and the resulting precipitate was collected and purified by chromatography (eluting with 168:15:1 CHCl3/MeOH/concentrated NH4OH) to supply 320 mg of the target 48 (80%) as a white solid judged by 1H NMR to be a 5.7:1 mixture of R,R/ minor diastereomers. 1H NMR (CDCl3) (R,R diastereomer): δ 1.11 (d, 3H, J ) 5.6 Hz), 2.27 (s, 3H), 2.73 (dd, 1H, J ) 8.4, 12.2 Hz), 2.84 (dd, 1H, J ) 3.6, 12.2 Hz), 2.93-2.97 (m, 2H), 3.11 (d, 1H, J ) 8.0 Hz), 4.31 (d, 2H, J ) 5.6 Hz), 4.40 (dd, 1H, J ) 3.6, 8.8 Hz), 5.68 (bs, 1H), 6.56 (d, 2H, J ) 8.4 Hz), 7.01-7.07 (m, 6H), 7.16-6.23 (m, 3H), 7.32 (s, 1H). HRMS ((M + H)+ found: 466.2250. Calcd for C27H32Cl1N3O2: 466.2261. Anal. (C27H32Cl1N3O2) C, H, N. tert-Butyl (2R)-2-{[tert-butyl(dimethyl)silyl]oxy}-2(3-chlorophenyl)ethyl{(1R)-1-methyl-2-[4-(2-{[(4-methylphenyl)sulfonyl]amino}ethyl)anilino]ethyl}carbamate (Intermediate 34). General procedure A was employed using aldehyde 10 (309 mg, 0.7 mmol), N-[2-(4-aminophenyl)ethyl]4-methylbenzenesulfonamide (19b, 163 mg, 0.56 mmol), NaBH(OAc)3 (262 mg, 1.24 mmol), and acetic acid (3 drops) in anhydrous CH2Cl2 (7.0 mL). Workup afforded 359 mg (90%) of intermediate 34 as a colorless glass. 1H NMR (CDCl3): δ -0.08 (s, 9H), 0.07 (s, 3H), 0.91 (m, 12H), 1.49 (s, 9H), 2.46 (s, 3H), 2.66 (t, 2H, J ) 6.9 Hz), 3.13-3.21 (m, 6H), 3.95-4.10 (m, 1H), 4.33 (m, 1H), 6.50 (d, 2H, J ) 8.4 Hz), 6.89 (d, 2H, J ) 8.4 Hz), 7.73 (d, 2H, J ) 8.1 Hz). MS (ES) m/e: 718.9, 715.8 (M + H).

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3 581 N-(2-{4-[((2R)-2-{[(2R)-2-Hydroxy-2-(3-chlorophenyl)ethyl]amino}propyl)amino]phenyl}ethyl)-4-methylbenzenesulfonamide (49). Intermediate 34 (359 mg, 0.50 mmol) was dissolved in 4 N HCl/dioxane (10 mL), and the mixture was stirred for 3 h. After removal of solvent, the resulting gum was purified by chromatography (eluting with 168:15:1 CHCl3/ MeOH/concentrated NH4OH) to supply 81.3 mg (32% yield) of the product 49 as a white foam judged by 1H NMR analysis to be a 5.3:1 mixture of R,R/minor diastereomers. 1H NMR (CDCl3) (R,R diastereomer): δ 1.11 (d, 3H, J ) 6.0 Hz), 2.38 (s, 3H), 2.59 (t, 2H, J ) 6.8 Hz), 2.72 (dd, 1H, J ) 8.8, 12.4 Hz), 2.84 (dd, 1H, J ) 3.6, 12.4 Hz), 2.90-2.98 (m, 3H), 3.003.12 (m, 4H), 4.40 (bs, 1H), 4.61 (dd, 1H, J ) 3.6, 8.8 Hz), 6.50 (d, 2H, J ) 8.4 Hz), 6.84 (d, 2H, J ) 8.4 Hz), 7.17-7.25 (m, 5H), 7.32 (s, 1H), 7.64 (d, 2H, J ) 8.0 Hz). HRMS ((M + H)+ found: 502.1949. Calcd for C26H32Cl1N3O3S1: 502.1931. Anal. (C26H32Cl1N3O3S1‚0.12CHCl3) C, H, N. Biological Methods. 1. In Vitro Functional Assays. In these experiments the β3 AR clone of Granneman and coworkers was employed.36 Chinese hampster ovary (CHO) cells expressing human β1, β2, or β3 AR were grown in DMEM/F12 (with pyroxidine‚HCl, 15 mM HEPES, l-glutamine), supplemented with 10% heat-inactivated FBS, 500 µg/mL G418, 2 mM L-glutamine, 100 units of penicillin G, and 100 µg of streptomycin sulfate. One confluent flask of cells was trypsinised and resuspended in the above medium at a concentration of 30-40000 cells/100 µL and plated into 96-well flat bottom plates. The cells were then used for assay within 1824 h. The medium was aspirated from each well and replaced with 180 µL of DMEM/F12 with 500 mM IBMX. The plate was then placed back in the incubator for 30 min. Drugs were then added to the wells (20 µL, 100 × required final concentration) for 60 min. Responses were determined by measuring cAMP levels of a 20 µL sample of extracellular media using a scintillation proximity based radioimmunoassay (NEN Flashplates). 2. Binding Assays. Human recombinant Sf9 cells expressing the cloned human β1 and β2 receptors were obtained using the method of Smith and Teitler.37 Receptor binding assays were carried out using the radioligand [125I]cyanopindolol at a concentration of 150 pM and the compound of interest at six concentrations ranging from 0.21 to 50 µM. Binding reactions were carried out for 1 h and 30 min for β1 and β2 receptors, respectively, at 22 °C and terminated by filtration through glass fiber filters (GF/B, Packard). Bound radioactivity was measured with a scintillation counter (Topcount, Packard) using a liquid scintillation cocktail (Microscint 0, Packard). 3. Pharmacokinetic Studies in Dogs. Male beagle dogs (weight range: 8-12 kg) were fasted overnight. On the morning of the study, each compound was dissolved in 0.025 M methanesulfonic acid containing 5% mannitol and the dogs were each fitted with two cephalic vein cannulae. Each dog was dosed with with one compound intravenously via one cannula at a dose level of 0.2 mg/kg body weight (5 min infusion period). Blood was collected via the second cephalic vein cannula at 0, 0.08, 0.25, 0.5, 0.75, 1, 1.5, 2.5, 4, 6, 8, and 24 h. Resulting serum samples were prepared by protein precipitation with acetonitrile and analyzed for compound content by APCI LC-MS/MS using a Hypersil BDS C18 column (30 mm × 1 mm, 3µm) at ambient temperature. A gradient mobile phase of 3-99% acetonitrile in 5 mM ammonium acetate buffer, pH 4.5, was used at a flow rate of 80 µL/min. Detection was by multiple reaction monitoring (MRM) on a Sciex API III+ with argon as the collision gas. Data reduction was performed using Sciex MacQuan software. Noncompartmental methods were used to calculate pharmacokinetic parameters from the resulting serum concentration vs time profiles. 4. Rodent Infrared Thermography Studies.35 CD-1 mice (CD-1(ICR)BR) were anesthetized with isoflurane and shaved to expose the interscapular region before IR imaging. The animals were orally dosed, and at the appropriate times they were anesthetized and scanned. Animals were dosed by oral gavage with either vehicle (0.025 M methanesulfoxide at 10

582

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3

mL/kg) or the test compound (10 mL/kg (volume), 1 mg/kg, 0.1 mg/kg, and 0.01 mg/kg (concentration)). The mice were placed in a manifold with nose ports for continual delivery of isoflurane. To maintain body core temperature during scanning, the rodents were placed on a tightly regulated heating table (37 ( 0.1 °C). The heating table was housed in an isothermal, nonreflective chamber (24 ( 0.1 °C, 50% relative humidity). Upon closure of the chamber door, heat emissions from the areas of interest were acquired using a highresolution InSb IR scanning detector (AGEMA Thermovision 900, Thermogenic Imaging, Billerica, MA) mounted 30 cm above the area of interest. Images were recorded at 1 min intervals for 5 min. A frame-averaging rate of 16 frames per second was used for each designated time point. Acquired images were analyzed for average temperatures using a GlaxoWellcome (RTP, NC) image-processing software application (RoboImage). Data were expressed as either average temperature per area or the change in temperature per area (drug treated minus vehicle treated). The data were calculated as the mean and standard error of the mean from experiments performed on 8-10 animals per treatment group. Two tailed t-tests were performed to calculate P values. Correlation coefficients were determined by regression analysis using Sigma Plot.

Acknowledgment. The authors thank Mike Martin for synthesizing intermediate 9 and Randy Rutkowski, Robert Johnson, and Peter Kitrinos for analytical support. The authors also gratefully acknowledge Mike Foxton, Rich Green, and Barry Shearer for helpful discussions. Supporting Information Available: HPLC traces of β3 agonists 35-49. This material is available free of charge via the Internet at http://pubs.acs.org.

Uehling et al.

(14)

(15)

(16)

(17) (18)

(19) (20)

References (1) Collins, P.; Williams, G. Drug treatment of obesity: from past failures to future sucesses? J. Clin. Pharmacol. 2001, 51, 1325. (2) Arch, J. R. S.; Ainsworth, A. T. Thermogenic and antiobesity activity of a novel β-adrenoceptor agonist (BRL 26830A) in mice and rats. Am. J. Clin. Nutr. 1983, 38, 549-558. (3) Arch, J. R. S.; Ainsworth, A. T.; Cawthorne, M. A.; Piercy, V.; Sennitt, M. V.; Thody, V. E.; Wilson, C.; Wilson, S. Atypical β-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature 1984, 309, 163-165. (4) Howe, R. β3-Adrenergic Agonists. Drugs Future 1993, 18, 529549. (5) Wilson, C.; Wilson, S.; Piercy, V.; Sennitt, M. V.; Arch, J. R. S. The rat lipolytic β-adrenoceptor: Studies using novel β-adrenoceptor agonists. Eur. J. Pharmacol. 1984, 100, 309-319. (6) Bloom, J. D.; Dutia, M. D.; Johnson, B. D.; Wissner, A.; Burns, M. G.; Largis, E. E.; Dolan, J. A.; Claus, T. H. Disodium (R,R)5-[2-[[2-(3-Chlorophenyl)-2-hydroxyethyl]-amino]propyl]-1,3-benzodioxole-2,2-dicarboxylate (CL 316243). A Potent β-adrenergic Agonist Virtually Specific for β3 Receptors. A Promising Antidiabetic and Antiobesity Agent. J. Med. Chem. 1992, 35, 30813084. (7) Lipworth, B. J. Clinical pharmacology of β3-adrenoceptors. Br. J. Clin. Pharmacol. 1996, 42, 291-300. (8) Weyer, C.; Tataranni, P. A.; Snitker, S.; Danforth, E., Jr.; Ravussin, E. Increase in Insulin Action and Fat Oxidation after Treatment with CL 316243, a Highly Selective β3-Adrenoceptor Agonist in Humans. Diabetes 1998, 47, 1555-1561. (9) Emorine, L. J.; Marullo, S.; Briend-Sutren, M.-M.; Patey, G.; Tate, K.; Delavier-Klutchko, C.; Strosberg, A. D. Molecular Characterization of the Human β3-Adrenergic Receptor. Science 1989, 245, 1118-1121. (10) Granneman, J. G.; Lahners, K. N.; Chaudhry, A. Molecular Cloning and Expression of the Rat β3-Adrenergic Receptor. Mol. Pharmacol. 1991, 40, 895-899. (11) Nahamias, C.; Blin, N.; Elalouf, J.-M.; Mattei, M. G.; Strosberg, A. D.; Emorine, L. J. Molecular characterization of the mouse β3-adrenergic receptor: relationship with the atypical receptor of adipocytes. EMBO J. 1991, 10, 3721-3727. (12) Strosberg, A. D.; Pietri-Rouxel, F. Function and regulation of the β3-adrenoceptor. Trends Pharmacol. Sci. 1996, 17, 373-381. (13) (a) Ghorbani, M.; Claus, T. H.; Himms-Hagen, J. Hypertrophy of Brown Adipocytes in Brown and White Adipose Tissues and Reversal of Diet-Induced Obesity in Rats Treated with a β3Adrenoceptor Agonist. Biochem. Pharmacol. 1997, 54, 121-131.

(21) (22)

(23) (24) (25)

(26)

(27) (28)

(29)

(b) Himms-Hagen, J.; Melnyk, A.; Zingaretti, M. C.; Ceresi, E.; Barbatelli, G.; Cinti, S. Multiocular fat cells in WAT of CL316,243-treated rats derive directly from white adipocytes. Am. J. Physiol. Cell Physiol. 2000, 279, C670-C681. Fisher, M. H.; Amend, A. M.; Bach, T. J.; Barker, J. M.; Brady, E. J.; Candelore, M. R.; Carrol, D.; Cascieri, M. A.; Chiu, S.-H. L.; Deng, L.; Forrest, M. J.; Hegarty-Friscino, B.; Guan, X.-M.; Hom, G. J.; Hutchins, J. E.; Kelly, L. J.; Mathvink, R. J.; Metzger, J. M.; Miller, R. R.; Ok, H. O.; Parmee, E. R.; Saperstein, R.; Strader, C. D.; Sterns, R. A.; Thompson, G. M.; Tota, L.; Vicario, P. P.; Weber, A. E.; Woods, J. W.; Wyvratt, M. J.; Zafian, P. T.; MacIntyre, D. E. A Selective Human β3 Adrenergic Receptor Agonist Increases Metabolic Rate in Rhesus Monkeys. J. Clin. Invest. 1998, 101, 2387-2393. Krief, S.; Lonnqvist, F.; Raimbault, S.; Baude, B.; Van Spronsen, A.; Arner, P.; Strosberg, A. D.; Ricquier, D.; Emorine, L. J. Tissue Distribution of β3-Adrenergic Receptor mRNA in Man. J. Clin. Invest. 1993, 91, 344-349. Chamberlain, P. D.; Jennings, K. H.; Paul, F.; Cordell, J.; Berry, A.; Holmes, S. D.; Park, J.; Chambers, J.; Sennitt, M. V.; Stock, M. J.; Cawthorne, M. A.; Young, P. W.; Murphy, G. J. The tissue distribution of the human beta(3)-adrenoceptor studied using amonoclonal antibody: Direct evidence of the beta(3)-adrenoceptor in human adipose tissue, atrium and skeletal muscle. Int. J. Obes. 1999, 23 (10), 1057-1065. Strosberg, A. D. Association of beta(3)-adrenoceptor polymorphism with obesity and diabetes. Trends Pharmacol. Sci. 1997, 18, 449-454. (a) Zheng, W.; Nikulin, V. I.; Konkar, A. A.; Vansal, S. S.; Shams, G.; Feller, D. R.; Miller, D. D. 2-Amino-4-benzyl-4,5,6,7-tetrahydrothiazolo[5,4-c]pyridines: Novel Selective β3-Adrenoceptor Agonists. J. Med. Chem. 1999, 42, 2287-2294. (b) Parmee, E. R.; Brockunier, L. L.; He, J.; Singh, S. B.; Candelore, M. R.; Cascieri, M. A.; Deng, L.; Liu, Y.; Tota, L.; Wyvratt, M. J.; Fisher, M. H.; Weber, A. E. Tetrahydroisoquinoline Derivatives Containing a Benzenesulfonamide Moiety as Potent, Selective Human β3 Adrenergic Receptor Agonists. Bioorg. Med. Chem. Lett. 2000, 10, 2283-2286. Weber, A. E. β3-Adrenergic receptor agonists for the treatment of obesity. Annu. Rep. Med. Chem. 1998, 33, 193-202. Mathvink, R. J.; Tolman, J. S.; Chitty, D.; Candelore, M. R.; Cascieri, M. A.; Colwell, L. F., Jr.; Deng, L.; Feeney, W. P.; Forrest, M. J.; Hom, G. J.; MacIntyre, D. E.; Miller, R. R.; Stearns, R. A.; Tota, L.; Wyvratt, M. J.; Fisher, M. H.; Weber, A. E. Discovery of a Potent, Orally Biovailable β3 Adrenergic Receptor Agonist, (R)-N-[4-[2-[[2-Hydroxy-2-(3-pyridinyl)ethyl]amino]ethyl]phenyl]-4-[4-[4-(trifluoromethyl)phenyl]thiazol-2-yl]benzenesulfonamide. J. Med. Chem. 2000, 43, 3832-3836. Howe, R.; Rao, B. S.; Holloway, B. R.; Stribling, D. J. Med. Chem. 1992, 35, 1751. (a) Sher, P. M.; Mathur, A.; Fisher, L. G.; Wu, G.; Skwish, S.; Michel, I. M.; Seiler, S. M.; Dickinson, K. E. Carboxyl-Promoted Enhancement of Selectivity for the β3 Adrenergic Receptor. Negative Charge of the Sulfonic Acid BMS-187413 Introduces β3 Binding Selectivity. Bioorg. Med. Chem. Lett. 1997, 7, 15831588. (b) Malamas, M. S.; Largis, E.; Gunawan, I.; Li, Z.; Tillett, J.; Han, S. C.-H.; Mulvey, R. Potent, Selective Aminothiazolidinediones Agonists of the Human β3 Adrenergic Receptor. Med. Chem. Res. 2000, 10, 164-177. Foxton, M. W. Patent WO 9533724, 1995. Additional acid isosteres in the aniline phenethanolamine series are described in the following. Hartley, C. D.; Carter, M. C.; Foxton, M. W. Patent WO 9721666, 1997. Khalaj, A.; Shadnia, H.; Sharifzadeh, M. Synthesis, molecular modeling and in vitro antibacterial activity of 4-amino-Rhydroxybenzeneacetamides. Pharm. Pharmacol. Commun. 1998, 4, 373-376. Sucholeiki, I.; Lynch, V.; Phan, L.; Wilcox, C. S. Chemistry of synthetic receptors and functional group arrays. 7. Molecular armatures. Synthesis and structure of Troeger’s base analogues derived from 4-, 2,4-, 3,4-, and 2,4,5-substituted aniline derivatives. J. Org. Chem. 1988, 53, 98-104. Drummond, J. T.; Johnson, G. Convenient Procedure for the Preparation of Alkyl and Aryl Substituted N-(Aminoalkylacyl)sulfonamides. Tetrahedron Lett. 1988, 29, 1653-1656. Clark, R. D.; Caroon, J. M.; Isaac, N. E.; McClelland, D. L.; Michel, A. D.; Petty, T. A.; Rosenkranz, R. P.; Waterbury, L. D. Synthesis and pharmacological evaluation of N,N-di-N-propyldopamine congeners containing phenolic bioisosteres. J. Pharm. Sci. 1987, 76, 411-415. Because they were not separable from the major diastereomers, the contribution of the minor R,S and S,R diastereomers to the profile of the target data presented in Tables 1 and 2 was not ascertained. However, investigations of the R,S, S,R, and S,S isomers of 1 have shown that R,S, S,R, and S,S diastereomers are substantially less active at all β AR subtypes than the R,R isomer. See the following reference. Oriowo, M. A.; Chapman, H.; Kirkham, D. M.; Sennit, M. V.; Ruffolo, R. R., Jr.; Cawthorne,

β3 Adrenergic Receptor Agonists M. A. J. Pharmacol. Exp. Ther. 1996, 277, 22. By analogy, we believe that the minor diastereomers of our compounds are substantially less active at all β AR subtypes and that their contribution to the profile of a mixture containing predominately R,R isomer is therefore negligible. See also ref 21, footnote 24, and references therein. (30) For examples of bioisosteric replacements of carboxylate with acylsulfonamide, sulfonylurea, or sulfonylsulfone in medicinal chemistry see the following. (a) Schuster, V. L.; Itoh, S.; Andrews, S. W.; Burk, R. M.; Chen, J.; Kedzie, K. M.; Gil, D. W.; Woodward, D. F. Synthetic Modification of Prostaglandin F2R Indicates Different Structural Determinants for Binding to the Prostaglandin F Receptor Versus the Prostaglandin Transporter. Mol. Pharmacol. 2000, 58, 1511-1516. (b) Deprez, P.; Guillaume, J.; Becker, R.; Corbier, A.; Didierlaurent, S.; Fortin, M.; Frechet, D.; Hamon, G.; Heckmann, B.; Heitsch, H.; Kleemann, H.-W.; Vevert, J.-P.; Vincent, J.-C.; Wagner, A.; Zhang, J. Sulfonylureas and Sulfonylcarbamates as New Non-Tetrazole Angiotensin II Receptor Antagonists. Discovery of a Highly Potent Orally Active (Imidazolylbiphenylyl)sulfonylurea (HR 720). J. Med. Chem. 1995, 38, 2357-2377. (c) Cunliffe, C. J.; Franklin, T. J.; Hales, N. J.; Hill, G. B. Novel Inhibitors of Prolyl 4-Hydroxylase. 3. Inhibition by the Substrate Analogue NOxaloglycine and Its Derivatives. J. Med. Chem. 1992, 35, 26522658. (31) Openshaw, T. T.; Spring, F. S. Preparation and properties of sulphonacetamides: a method for the separation of sulfonamides from N-alkylsulfonamides. J. Chem. Soc. 1945, 234-236. (32) For recent examples, see the following. (a) Feng, D. D.; Biftu, T.; Candelore, M. R.; Cascieri, L. F., Jr.; Deng, L.; Feeney, W. P.; Forrest, M. J.; Hom, G. J.; MacIntyre, D. E.; Miller, R. R.;

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 3 583

(33)

(34)

(35) (36) (37)

Stearns, R. A.; Strader, C. D.; Tota, L.; Wyvratt, M. J.; Fisher, M. H.; Weber, A. E. Discovery of an Orally Bioavailable Alkyl Oxadiazole β3 Adrenergic Receptor Agonist. Bioorg. Med. Chem. Lett. 2000, 10, 1427-1429. (b) Shih, T. L.; Candelore, M. R.; Cascieri, M. A.; Shuet-Hing, L.; Chiu, S.-H. L.; Colwell, L. F., Jr.; Deng, L.; Feeney, W. P.; Forrest, M. J.; Hom, G. J.; MacIntyre, D. E.; Miller, R. R.; Stearns, R. A.; Strader, C. D.; Tota, L.; Wyvratt, M. J.; Fisher, M. H.; Weber, A. E. L-770,644: A Potent and Selective Human β3 Adrenergic Receptor Agonist with Improved Oral Bioavailability. Bioorg. Med. Chem. Lett. 1999, 1251-1254. For examples of rapidly cleaved acylsulfonamides see the following. Talley, J. J.; Bertenshaw, S. R.; Brown, D. L.; Carter, J. S.; Graneto, M. J.; Kellogg, M. S.; Koboldt, C. M.; Yuan, J.; Zhang, Y. Y.; Seibert, K. N-[[(5-Methyl-3-phenylisoxazole-4-yl)phenyl]sulfonyl]propanamide, Sodium Salt, Parecoxib Sodium: A Potent and Selective Inhibitor of COX-2 for Parenteral Administration. J. Med. Chem. 2000, 43, 1661-1663. Larsen, J. D.; Bundgaard, H. Prodrug forms for the sulfonamide group. I. Evaluation of N-acyl derivatives, N-sulfonylamidines, N-sulfonylsulfilimines and sulfonylureas as possible prodrug derivatives. Int. J. Pharm. 1987, 37, 87-95. Paulik, M. A.; Rose, M. L.; Lancaster, M. E.; Spencer, D. H.; Miller, R. T.; Weiel, J. E.; Lenhard, J. M. J. Pharm. Sci., in press. Granneman, J. G.; Lahners, K. N.; Rao, D. D. Mol. Pharmacol. 1992, 42, 964. Smith, C.; Teitler, M. Cardiovasc. Drugs Ther. 1999, 13, 123-126.

JM0101500